Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Am J Sports Med ; 52(2): 362-367, 2024 02.
Article in English | MEDLINE | ID: mdl-38230875

ABSTRACT

BACKGROUND: Costal chondrocyte-derived pellet-type autologous chondrocyte implantation (CCP-ACI) has been introduced as a new therapeutic option for the treatment of articular cartilage defects. We had previously conducted a randomized controlled trial comparing CCP-ACI versus microfracture at 1 year postoperatively. PURPOSE: To compare the efficacy and safety of CCP-ACI versus microfracture for the treatment of articular cartilage defects of the knee at 5 years postoperatively. STUDY DESIGN: Randomized controlled trial; Level of evidence, 2. METHODS: This study describes the mean 5-year follow-up of a previously published prospective clinical trial. The previous prospective trial compared the results of CCP-ACI versus microfracture until 1 year of follow-up. Of the 30 patients who were included in the previous study, 25 were followed up for 5 years. Patients were evaluated based on clinical outcome scores (Lysholm score, International Knee Documentation Committee score, Knee injury and Osteoarthritis Outcome Score [KOOS], and visual analog scale for pain), magnetic resonance imaging findings, and rates of treatment failure at last follow-up. RESULTS: The MOCART (Magnetic Resonance Observation of Cartilage Repair Tissue) score in the CCP-ACI group was significantly higher than that in the microfracture group at 5 years (62.3 vs 26.7, respectively; P < .0001). The Lysholm score and KOOS score in the CCP-ACI group were significantly higher than those in the microfracture group at 5 years (84.5 vs 64.9, respectively, and 390.9 vs 303.0, respectively; P = .023 and P = .017, respectively). There was 1 case of treatment failure that occurred in the microfracture group. CONCLUSION: The present randomized controlled trial indicated that the results of both procedures clinically and statistically significantly improved at 1 and 5 years' follow-up in treating cartilage defects, but the results of CCP-ACI were superior to those of microfracture. Magnetic resonance imaging conducted at 1 year and 5 years after CCP-ACI revealed statistically significant superior structural integration with native cartilage tissue compared with microfracture. REGISTRATION: NCT03545269 (ClinicalTrials.gov).


Subject(s)
Cartilage, Articular , Fractures, Stress , Humans , Cartilage, Articular/injuries , Chondrocytes/transplantation , Follow-Up Studies , Fractures, Stress/pathology , Prospective Studies , Transplantation, Autologous/methods , Knee Joint/surgery
2.
Methods Mol Biol ; 2598: 65-73, 2023.
Article in English | MEDLINE | ID: mdl-36355285

ABSTRACT

Bone marrow-derived mesenchymal stromal cells (BM-MSC) are widely studied in the field of cartilage regeneration due to their capacity to differentiate into chondrocytes under specific in vitro culture conditions. This chapter describes the isolation of MSC from bone marrow aspirate, their expansion in monolayer, and the chondrogenic differentiation in pellet culture.


Subject(s)
Bone Marrow , Mesenchymal Stem Cells , Humans , Bone Marrow Cells , Chondrogenesis , Cell Differentiation , Chondrocytes , Cells, Cultured
3.
Front Bioeng Biotechnol ; 10: 971932, 2022.
Article in English | MEDLINE | ID: mdl-36561039

ABSTRACT

This study examined the role of sirtuins in the regenerative potential of articular chondrocytes. Sirtuins (SIRT1-7) play a key role in regulating cartilage homeostasis. By inhibiting pro-inflammatory pathways responsible for cartilage degradation and promoting the expression of key matrix components, sirtuins have the potential to drive a favourable balance between anabolic and catabolic processes critical to regenerative medicine. When subjected to osmolarity and glucose concentrations representative of the in vivo niche, freshly isolated bovine chondrocytes exhibited increases in SIRT1 but not SIRT3 gene expression. Replicating methods adopted for the in vitro monolayer expansion of chondrocytes for cartilage regenerative therapies, we found that SIRT1 gene expression declined during expansion. Manipulation of sirtuin activity during in vitro expansion by supplementation with the SIRT1-specific activator SRT1720, nicotinamide mononucleotide, or the pan-sirtuin inhibitor nicotinamide, significantly influenced cartilage regeneration in subsequent 3D culture. Tissue mass, cellularity and extracellular matrix content were reduced in response to sirtuin inhibition during expansion, whilst sirtuin activation enhanced these measures of cartilage tissue regeneration. Modulation of sirtuin activity during monolayer expansion influenced H3K27me3, a heterochromatin mark with an important role in development and differentiation. Unexpectedly, treatment of primary chondrocytes with sirtuin activators in 3D culture reduced their matrix synthesis. Thus, modulating sirtuin activity during the in vitro monolayer expansion phase may represent a distinct opportunity to enhance the outcome of cartilage regenerative medicine techniques.

4.
Tissue Eng Part C Methods ; 28(10): 501-510, 2022 10.
Article in English | MEDLINE | ID: mdl-36082992

ABSTRACT

Transforming growth factor-beta 1 (TGF-ß1) has been reported to promote chondrogenic differentiation and proliferation in the multipotent stromal cell (MSCs), and the transforming growth factor-beta 3 (TGF-ß3) tends to be exclusively in promoting cell differentiation alone. The objective of this study was to determine the effect of TGF-ß1 and -ß3 on the MSCs chondrogenic differentiation on the poly (vinyl alcohol)-chitosan-poly (ethylene glycol) (PVA-NOCC-PEG) scaffold, compared with that of monolayer and pellet cultures. In this study, P2 rabbit bone marrow-derived MSCs were seeded either on the untreated six-well plate (for monolayer culture) or onto the PVA-NOCC-PEG scaffold or cultured as a pellet culture. The cultures were maintained in a chemically defined serum-free medium supplemented with 10 ng/mL of either TGF-ß1 or TGF-ß3. Cell viability assay, biochemical assay, and real-time polymerase chain reaction were performed to determine the net effect of cell proliferation and chondrogenic differentiation of each of the growth factors. The results showed that the PVA-NOCC-PEG scaffold enhanced MSCs cell proliferation from day 12 to 30 (p < 0.05); however, no significant differences were observed in the cell proliferation between the cultures supplemented with or without TGF-ß1 and TGF-ß3 (p > 0.05). In terms of chondrogenic differentiation, the PVA-NOCC-PEG scaffold augmented the GAGs secretion in MSCs and the mRNA expression levels of Sox9, Col2a1, Acan, and Comp were elevated (p < 0.05). However, there was no significant difference between both the TGF-ß1 and TGF-ß3-treated groups (p > 0.05). In conclusion, TGF-ß1 and TGF-ß3 enhanced the chondrogenic differentiation of MSCs seeded on the PVA-NOCC-PEG scaffold; however, there was no significant difference between the effect of TGF-ß1 and TGF-ß3. Impact statement Transforming growth factor-beta (TGF-ß) superfamily members is a key requirement for the in vitro chondrogenic differentiation of mesenchymal stem cells (MSCs). In this study, the effects of TGF-ß1 and -ß3 on MSC chondrogenic differentiation and proliferation on a novel three-dimensional scaffold, the poly(vinyl alcohol)-chitosan-poly(ethylene glycol) (PVA-NOCC-PEG) scaffold, was evaluated. In this study, the results showed both TGF-ß1 and TGF-ß3 can enhance the chondrogenic differentiation of MSCs seeded on the PVA-NOCC-PEG scaffold.


Subject(s)
Chitosan , Mesenchymal Stem Cells , Animals , Rabbits , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/pharmacology , Polyvinyl Alcohol/pharmacology , Polyvinyl Alcohol/metabolism , Chitosan/pharmacology , Chitosan/metabolism , Transforming Growth Factor beta1/pharmacology , Polyethylene Glycols/pharmacology , Chondrogenesis , Cell Differentiation , Transforming Growth Factor beta/pharmacology , Transforming Growth Factors/metabolism , Transforming Growth Factors/pharmacology , Cells, Cultured
5.
Curr Issues Mol Biol ; 43(2): 665-675, 2021 Jul 11.
Article in English | MEDLINE | ID: mdl-34287259

ABSTRACT

Intra-articular injection of different types of blood-derived products is gaining popularity and clinical importance in the treatment of degenerative cartilage disorders such as osteoarthritis. The regenerative potential of two types of platelet-rich plasma (PRP), prepared in the presence of EDTA (EPRP) and citrate (CPRP) and an alternative blood product-hyperacute serum (hypACT) was evaluated using a 3D osteoarthritic chondrocyte pellet model by assessing the metabolic cell activity, cartilage-related gene expression and extracellular matrix deposition within the pellets. Chondrocyte viability was determined by XTT assay and it revealed no significant difference in metabolic activity of OA chondrocyte pellets after supplementation with different blood products. Nevertheless, the selection of blood products influenced the cartilage-related genes expression, ECM morphology and the tissue quality of pellets. Both PRP types had a different biological effect depending upon concentration and even though CPRP is widely used in clinics our assessment did not reveal good results in gene expression either tissue quality. HypACT supplementation resulted in superior cartilage-related genes expression together with tissue quality and seemed to be the most stable product since no remarkable changes were observed between the two different concentrations. All in all, for successful regenerative therapy, possible molecular mechanisms induced by blood-derived products should be always carefully investigated and adapted to the specific medical indications.


Subject(s)
Chondrocytes/cytology , Chondrocytes/drug effects , Osteogenesis , Platelet-Rich Fibrin , Platelet-Rich Plasma , Regeneration , Adult , Biomarkers , Cell Culture Techniques , Cells, Cultured , Chondrocytes/metabolism , Energy Metabolism , Extracellular Matrix/metabolism , Female , Gene Expression Regulation , Humans , Immunohistochemistry , Male , Middle Aged , Osteoarthritis/etiology , Osteoarthritis/metabolism , Osteoarthritis/therapy , Platelet-Rich Fibrin/metabolism , Platelet-Rich Plasma/metabolism
6.
Vet J ; 269: 105605, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33593496

ABSTRACT

Mesenchymal stromal cells (MSC) are used for cell-based treatment for canine osteoarthritis (OA). Compared with human MSCs, detailed information on the functional characterisation of canine MSCs is limited. In particular, the chondrogenic differentiation of canine adipose tissue-derived MSCs (cAT-MSCs) is challenging. In this study, we aimed to compare cAT-MSCs with bone marrow-derived MSCs (cBM-MSCs), focusing specifically on their in vitro chondrogenic potential, with or without bone morphogenetic proteins (BMP). cBM-MSCs and cAT-MSCs were characterised using flow cytometry and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The chondrogenic differentiation potential of all cMSC preparations in the presence of TGF-ß1 alone or when supplemented with 10, 100, or 250 ng/mL BMP-2 or BMP-6 was investigated using RT-qPCR, and biochemical, histochemical and immunohistological analyses. Both cBM-MSCs and cAT-MSCs expressed the surface markers CD90, CD73, and CD29, and were negative for CD45 and CD34, although the expression of CD73 and CD271 varied with donor and tissue origin. Interestingly, expression of ACAN and SOX9 was higher in cBM-MSCs than cAT-MSCs. In contrast with cBM-MSCs, cAT-MSCs could not differentiate toward the chondrogenic lineage without BMP-2/-6, and their in vitro chondrogenesis was inferior to cBM-MSCs with BMP-2/-6. Thus, cAT-MSCs have lower in vitro chondrogenic capacity than cBM-MSC under the studied culture conditions with 10, 100, or 250 ng/mL BMP-2 or BMP-6. Therefore, further characterisation is necessary to explore the potential of cAT-MSCs for cell-based OA treatments.


Subject(s)
Bone Marrow Cells/physiology , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 6/pharmacology , Chondrogenesis/physiology , Mesenchymal Stem Cells/physiology , Animals , Antigens, Surface/analysis , Cell Culture Techniques/veterinary , Cell Differentiation/drug effects , Colony-Forming Units Assay/veterinary , Dog Diseases/therapy , Dogs , Mesenchymal Stem Cell Transplantation , Osteoarthritis/therapy , Osteoarthritis/veterinary , Transforming Growth Factor beta1/pharmacology
7.
Biofabrication ; 13(3)2021 04 07.
Article in English | MEDLINE | ID: mdl-33513590

ABSTRACT

Scaffold-free 3D cell cultures (e.g. pellet cultures) are widely used in medical science, including cartilage regeneration. Their drawbacks are high time/reagent consumption and lack of early readout parameters. While optimisation was achieved by automation or simplified spheroid generation, most culture systems remain expensive or require tedious procedures. The aim of this study was to establish a system for resource efficient spheroid generation with additional early readout parameters. This was achieved by a new approach for spheroid generation via self-assembly from monolayer via compartmentation of cell culture surfaces utilising laser engraving (grid plates). The compartmentation triggered contraction and rolling up of the cell monolayer, finishing in condensation into a spheroid in human adipose-derived stem cell (ASC/TERT1) and human articular chondrocytes (hACs)-ASC/TERT1 co-cultures, when cultivated on grid plates under chondrogenic conditions. Plates with 1 and 3 mm grid size yielded stable diameters (about 140µm and 300µm, respectively). ASC/TERT1 spheroids fully formed within 3 weeks while co-cultures took 1-2 weeks, forming significantly faster with increasing hAC ratio (p< 0.05 and 0.01 for 1:1 and 1:4 ASC/TERT1:hAC ratio, respectively). Co-cultures showed slightly lower spheroid diameters, due to earlier spheroid formation and incomplete monolayer formation. However, this was associated with a more homogeneous matrix distribution in the co-culture. Both showed differentiation capacity comparable to standard pellet culture in (immune-)histochemistry and RT-qPCR. To assess usability for cartilage repair, spheroids were embedded into a hydrogel (fibrin), yielding cellular outgrowth and matrix deposition, which was especially pronounced in co-cultures. The herein presented novel cell culture system is not only a promising tool for autonomous spheroid generation with the potential of experimental and clinical application in tissue engineering, but also for the generation of 'building blocks' for subsequential biofabrication strategies such as bioprinting.


Subject(s)
Cell Culture Techniques, Three Dimensional , Chondrogenesis , Cell Differentiation , Cells, Cultured , Chondrocytes , Humans , Tissue Engineering
8.
Front Cell Dev Biol ; 8: 279, 2020.
Article in English | MEDLINE | ID: mdl-32500073

ABSTRACT

Human dental pulp stem cells (hDPSCs) are characterized by high proliferation rate, the multi-differentiation ability and, notably, low immunogenicity and immunomodulatory properties exerted through different mechanisms including Fas/FasL pathway. Despite their multipotency, hDPSCs require particular conditions to achieve chondrogenic differentiation. This might be due to the perivascular localization and the expression of angiogenic marker under standard culture conditions. FasL stimulation was able to promote the early induction of chondrogenic commitment and to lead the differentiation at later times. Interestingly, the expression of angiogenic marker was reduced by FasL stimulation without activating the extrinsic apoptotic pathway in standard culture conditions. In conclusion, these findings highlight the peculiar embryological origin of hDPSCs and provide further insights on their biological properties. Therefore, Fas/FasL pathway not only is involved in determining the immunomodulatory properties, but also is implicated in supporting the chondrogenic commitment of hDPSCs.

9.
Article in English | MEDLINE | ID: mdl-32432090

ABSTRACT

Three-dimensional (3D) cell spheroids are being increasingly applied in many research fields due to their enhanced biological functions as compared to conventional two-dimensional (2D) cultures. 3D cell spheroids can replicate tissue functions, which enables their use both as in vitro models and as building blocks in tissue biofabrication approaches. In this study, we developed a perfusable microfluidic platform suitable for robust and reproducible 3D cell spheroid formation and tissue maturation. The geometry of the device was optimized through computational fluid dynamic (CFD) simulations to improve cell trapping. Experimental data were used in turn to generate a model able to predict the number of trapped cells as a function of cell concentration, flow rate, and seeding time. We demonstrated that tuning non-geometrical parameters it is possible to control the size and shape of 3D cell spheroids generated using articular chondrocytes (ACs) as cellular model. After seeding, cells were cultured under perfusion at different flow rates (20, 100, and 500 µl/min), which induced the formation of conical and spherical spheroids. Wall shear stress values on cell spheroids, computed by CFD simulations, increased accordingly to the flow rate while remaining under the chondroprotective threshold in all configurations. The effect of flow rate on cell number, metabolic activity, and tissue-specific matrix deposition was evaluated and correlated with fluid velocity and shear stress distribution. The obtained results demonstrated that our device represents a helpful tool to generate stable 3D cell spheroids which can find application both to develop advanced in vitro models for the study of physio-pathological tissue maturation mechanisms and to obtain building blocks for the biofabrication of macrotissues.

10.
Tissue Eng Regen Med ; 17(2): 165-181, 2020 04.
Article in English | MEDLINE | ID: mdl-32193874

ABSTRACT

BACKGROUND: To regenerate tissue-engineered cartilage as a source of material for the restoration of cartilage defects, we used a human fetal cartilage progenitor cell pellet to improve chondrogenesis and modulation of the immune response in an in vivo bioreactor (IVB) system. METHODS: IVB was buried subcutaneously in the host and then implanted into a cartilage defect. The IVB was composed of a silicone tube and a cellulose nano pore-sized membrane. First, fetal cartilage progenitor cell pellets were cultured in vitro for 3 days, then cultured in vitro, subcutaneously, and in an IVB for 3 weeks. First, the components and liquidity of IVB fluid were evaluated, then the chondrogenesis and immunogenicity of the pellets were evaluated using gross observation, cell viability assays, histology, biochemical analysis, RT-PCR, and Western blots. Finally, cartilage repair and synovial inflammation were evaluated histologically. RESULTS: The fluid color and transparency of the IVB were similar to synovial fluid (SF) and the components were closer to SF than serum. The IVB system not only promoted the synthesis of cartilage matrix and maintained the cartilage phenotype, it also delayed calcification compared to the subcutaneously implanted pellets. CONCLUSION: The IVB adopted to study cell differentiation was effective in preventing host immune rejection.


Subject(s)
Bioreactors , Cellulose/chemistry , Chondrogenesis , Immunity , Tissue Engineering , Animals , Cartilage, Articular , Cell Differentiation , Cell Survival , Chondrocytes/cytology , Humans , Male , Mesenchymal Stem Cells/cytology , Rabbits , Stem Cells , Synovial Fluid
11.
Molecules ; 24(20)2019 Oct 13.
Article in English | MEDLINE | ID: mdl-31614911

ABSTRACT

Combinations of IL-1ß and other proinflammatory cytokines reportedly promote the severity of arthritis. We aimed to investigate the effects of IL-1ß combined with IL-17A on cartilage degradation and synthesis in in vitro models. Cartilage explant degradation was determined using sulfated glycosaminoglycans (S-GAGs) levels, matrix metalloproteinase (MMP13) gene expression, uronic acid, and collagen contents. Cell morphology and accumulation of proteoglycans were evaluated using hematoxylin-eosin and safranin O staining, respectively. In the pellet culture model, expressions of cartilage-specific anabolic and catabolic genes were evaluated using real-time qRT-PCR. Early induction of MMP13 gene expression was found concomitantly with significant S-GAGs release. During the prolonged period, S-GAGs release was significantly elevated, while MMP-13 enzyme levels were persistently increased together with the reduction of the cartilaginous matrix molecules. The pellet culture showed anabolic gene downregulation, while expression of the proinflammatory cytokines, mediators, and MMP13 genes were elevated. After cytokine removal, these effects were restored to nearly basal levels. This study provides evidence that IL-1ß combined with IL-17A promoted chronic inflammatory arthritis by activating the catabolic processes accompanied with the suppression of cartilage anabolism. These suggest that further applications, which suppress inflammatory enhancers, especially IL-17A, should be considered as a target for arthritis research and therapy.


Subject(s)
Arthritis, Rheumatoid/genetics , Interleukin-17/genetics , Interleukin-1beta/genetics , Matrix Metalloproteinase 13/genetics , Animals , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cartilage, Articular/growth & development , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrogenesis/genetics , Gene Expression Regulation , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Humans , Inflammation/genetics , Inflammation/pathology , Metabolism , Proteoglycans/genetics , Swine , Tumor Necrosis Factor-alpha/genetics
12.
BMC Res Notes ; 12(1): 16, 2019 Jan 14.
Article in English | MEDLINE | ID: mdl-30642366

ABSTRACT

OBJECTIVE: This study aimed to clarify the effects of periodic mild heat stimuli on extracellular matrix (ECM) synthesis of adult human chondrocytes in 3-dimensional pellet culture. RESULTS: Human articular chondrocytes were subjected to pellet culture at 37 °C for 3 days. Thereafter, the pellets were divided into three groups: 32 °C group which was cultured at 32 °C without heat stimuli, 32 °C + Heat group which was cultured at 32 °C and applied periodic heat stimuli, 37 °C group which was cultured at 37 °C. Heat stimuli were given by transferring the pellets into a CO2 incubator set at 41 °C for 20 min/day, 6 times/week. ECM synthesis ability was evaluated by analyzing the mRNA expressions. Additionally, the collagen and proteoglycan content in the pellet was quantified. DNA content was also measured for estimating the cell amount. We found that there were no significant differences in the mRNA expression of COL2A1, COL1A1, and ACAN between the 32 °C group and 32 °C + Heat group. However, the collagen content per cell and DNA content were significantly lower in the 32 °C + Heat group compared to other groups. Our results indicate that periodic mild heat stimuli may diminish ECM synthesis due to inhibition of collagen production and loss of cells.


Subject(s)
Aggrecans/metabolism , Chondrocytes/metabolism , Collagen Type II/metabolism , Collagen Type I/metabolism , Extracellular Matrix/metabolism , Hot Temperature , Cells, Cultured , Collagen Type I, alpha 1 Chain , Humans
13.
Adv Pharm Bull ; 8(2): 283-289, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30023330

ABSTRACT

Purpose: In Persian traditional medicine, application of Mummy material has been advised since hundred years ago for treatment of different diseases as bone fracture, cutaneous wounds and joint inflammation. Regarding to the claim of indigenous people for application of this material in the treatment of joint inflammation, the present study was designed to evaluate whether Mummy can revoke the inflammatory responses in chondrocytes stimulated with interleukin 1-ß (IL-1ß). Methods: Isolated chondrocytes at the second passage were plated in 50 ml conical tubes at density of 1x106 for pellet culture or were plated in T75 culture flasks as monolayer. Cells in both groups were treated as control (receiving serum free culture medium), negative control (receiving IL-1ß (10ng/ml for 24 hr)) and IL-1ß pre-stimulated cells which treated with Mummy at concentrations of 500 and 1000µg/ml for 72hrs. After 72 hrs, to evaluate whether Mummy can revoke the inflammatory response in chondrocytes, cell in different groups were prepared for investigation of gene expression profile of collagen II, Cox-2, MMP-13, C-Rel and P65 using real-time RT-PCR. Results: Treatment of chondrocytes with IL-1ß (10ng/ml) resulted in a significant increase in expression level of Cox-2, MMP-13, C-Rel and P65 in pellet culture system, while treatment of IL-1ß-stimulated choncrocytes with Mummy at both concentrations of 500 and 1000µg/ml inhibited the expression level of above mentioned genes. Compared to the pellet culture, Mummy did not affect expression level of genes in monolayer condition. Conclusion: The obtained data from this investigation revealed that Mummy can be used as a potent factor for inhibiting the inflammatory responses induced by IL-1ß in chondrocytes probably through inhibition of NF-қB subunits activation.

14.
Arch Oral Biol ; 91: 1-8, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29621667

ABSTRACT

OBJECTIVE: Three-dimensional (3D) cell culture methods are of high importance to studies of biological processes. This is particularly the case with spheroid cultures, which create 3D cell aggregates without the use of exogenous materials. Compared to conventional monolayer cultures, cellular spheroid cultures have been demonstrated to improve multilineage potential and extracellular matrix production. To address this issue in depth, we present a more comprehensive analysis of 3D human dental pulp cell (hDPC) spheroids. DESIGN: hDPC spheroids were fabricated by the pellet culture method and were cultured without adding any reagent to induce differentiation. The gene-expression profiles of the 3D and two-dimensional (2D) cultured hDPCs were compared by complementary DNA microarray analysis. Odontoblastic and osteoblastic differentiation marker gene expression was evaluated by quantitative real-time PCR (RT-qPCR). Hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM) were applied to examine the morphology of hDPC spheroids and extracellular matrix components. RESULTS: Compared with 2D monolayer culture, microarray analysis identified 405 genes and 279 genes with twofold or greater differential expression after 3 days and 28 days of 3D culture, respectively. In 3D hDPC spheroids, gene ontology analysis revealed upregulation of extracellular matrix-related genes and downregulation of cell growth-related genes. RT-qPCR analysis showed higher expression levels of osteocalcin, dentin sialophosphoprotein, and alkaline phosphatase. TEM revealed the morphological characteristics of the fibrillar collagen-rich matrix and cell-cell interactions. CONCLUSIONS: The present findings provide clues to understanding the mechanisms of pellet-cultured hDPCs and contribute to future research in the comparative studies of different 3D culture methods.


Subject(s)
Cell Culture Techniques/methods , Dental Pulp/cytology , Dental Pulp/physiology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Cells, Cultured , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Microscopy, Electron, Transmission , Odontoblasts/cytology , Osteocalcin/genetics , Osteocalcin/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Transcriptome
15.
BMC Musculoskelet Disord ; 18(1): 432, 2017 Nov 06.
Article in English | MEDLINE | ID: mdl-29110708

ABSTRACT

BACKGROUND: The aim was to investigate whether resident chondrocytes in human articular cartilage and in subculture express vitamin D receptor (VDR) and the enzyme that hydroxylates the prohormone 25(OH)D3 to the active hormone 1α,25(OH)2D3, namely 1α-hydroxylase (CYP27B1). Any putative effects of vitamin D on chondrocytes were also explored. METHODS: Cartilage from human osteoarthritic knee joints, cultured chondrocytes and cells grown in 3D spheroids were examined for the expression of VDR and 1α-hydroxylase by PCR, Western blots and immunolabelling. Receptor engagement was judged by visualizing nuclear translocation. The effects of 25(OH)D3 and 1α,25(OH)2D3 on chondrocyte functions were assessed in proliferation-, chondrogenesis- and cartilage signature-gene expression assays. The capability of chondrocytes to hydroxylate 25(OH)D3 was determined by measuring the concentration of metabolites. Finally, a putative regulation of receptor and enzyme expression by 1α,25(OH)2D3 or interleukin (IL)-1ß, was investigated by Western blot. RESULTS: Gene expression was positive for VDR in freshly isolated cells from native cartilage, cells subcultured in monolayers and in spheroids, whereas protein expression, otherwise judged low, was apparent in monolayers. Nuclear translocation of VDR occurred upon 1α,25(OH)2D3 treatment. Transcripts for 1α-hydroxylase were detected in freshly isolated cells, cultured cells and spheroids. Western blots and immunolabelling detected 1α-hydroxylase protein in all materials, while staining of tissue appeared confined to cells at the superficial layer. A dose-dependent 1α,25(OH)2D3 production was measured when the enzyme substrate was supplied to cell cultures. Western blots revealed that the VDR, but not 1α-hydroxylase, was induced by IL-1ß treatment in adherent cells. Proliferation in monolayers was enhanced by both 25(OH)D3 and 1α,25(OH)2D3, and both compounds had negative effects on chondrogenesis and cartilage-matrix genes. CONCLUSIONS: VDR expression in resident cartilage chondrocytes, generally considered differentiated cells, is elusive. A similar pattern applies for redifferentiated chondrocytes in spheroid cultures, whereas dedifferentiated cells, established in monolayers, stably express VDR. Both 25(OH)D3 and 1α,25(OH)2D3 are able to potentiate cell proliferation but have a negative impact in proteoglycan synthesis. Chondrocytes express 1α-hydroxylase and may contribute to the production of 1α,25(OH)2D3 into the joint environment. Effects of vitamin D could be unfavourable in the context of cartilage matrix synthesis.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Chondrocytes/enzymology , Receptors, Calcitriol/metabolism , Adult , Aged , Cartilage, Articular/cytology , Cell Dedifferentiation , Cellular Reprogramming , Chondrogenesis , Humans , Middle Aged , Osteoarthritis, Knee/enzymology , Primary Cell Culture
16.
J Tissue Eng ; 8: 2041731417732655, 2017.
Article in English | MEDLINE | ID: mdl-29051809

ABSTRACT

A critical limitation in the cultivation of cartilage for tissue engineering is the dedifferentiation in chondrocytes, mainly during in vitro amplification. Despite many previous studies investigating the influence of various conditions, no data exist concerning the effects of hypothermia. Our aim has been to influence chondrocyte dedifferentiation in vitro by hypothermic conditions. Chondrocytes were isolated from cartilage biopsies and seeded in monolayer and in three-dimensional pellet-cultures. Each cell culture was either performed at 32.2°C or 37°C during amplification. Additionally, the influence of the redifferentiation of chondrocytes in three-dimensional cell culture was examined at 32.2°C and 37°C after amplification at 32.2°C or 37°C. An 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay was used to measure cell proliferation in monolayer, whereas the polymerase chain reaction and immunohistochemical and histological staining were used in three-dimensional pellet-cultures. Real-time polymerase chain reaction was employed to measure the relative expression of the target genes collagen II, collagen I, aggrecan and versican. Ratios were estimated between collagen II/collagen I and aggrecan/versican to evaluate differentiation. A higher value of these ratios indicated an advantageous status of differentiation. In monolayer, hypothermia at 32.2°C slowed down the proliferation rate of chondrocytes significantly, being up to two times lower at 32.2°C compared with culture at 37°C. Simultaneously, hypothermia in monolayer decelerated dedifferentiation. The ratio of aggrecan/versican was significantly higher at 32.2°C compared with that at 37°C. In three-dimensional pellet-culture, the chondrocytes redifferentiated at 32.2°C and at 37°C, and this process is more distinct at 37°C than at 32.2°C. Similar results were obtained for the ratios of collagen II/collagen I and aggrecan/versican and were supported by immunochemical and histological staining. Thus, hypothermic conditions for chondrocytes are mainly advantageous in monolayer culture. In three-dimensional pellet-culture, redifferentiation predominates at 37°C compared with at 32.2°C. In particular, the results from the monolayer cultures show potential in the avoidance of dedifferentiation.

17.
Front Pharmacol ; 8: 255, 2017.
Article in English | MEDLINE | ID: mdl-28536528

ABSTRACT

Background: Mesenchymal stem cells (MSCs) and their chondrogenic differentiation have been extensively investigated in vitro as MSCs provide an attractive source besides chondrocytes for cartilage repair therapies. Here we established prototype foamyviral vectors (FVV) that are derived from apathogenic parent viruses and are characterized by a broad host range and a favorable integration pattern into the cellular genome. As the inflammatory cytokine interleukin 1 beta (IL1ß) is frequently present in diseased joints, the protective effects of FVV expressing the human interleukin 1 receptor antagonist protein (IL1RA) were studied in an established in vitro model (aggregate culture system) of chondrogenesis in the presence of IL1ß. Materials and Methods: We generated different recombinant FVVs encoding enhanced green fluorescent protein (EGFP) or IL1RA and examined their transduction efficiencies and transgene expression profiles using different cell lines and human primary MSCs derived from bone marrow-aspirates. Transgene expression was evaluated by fluorescence microscopy (EGFP), flow cytometry (EGFP), and ELISA (IL1RA). For evaluation of the functionality of the IL1RA transgene to block the inhibitory effects of IL1ß on chondrogenesis of primary MSCs and an immortalized MSC cell line (TERT4 cells), the cells were maintained following transduction as aggregate cultures in standard chondrogenic media in the presence or absence of IL1ß. After 3 weeks of culture, pellets were harvested and analyzed by histology and immunohistochemistry for chondrogenic phenotypes. Results: The different FVV efficiently transduced cell lines as well as primary MSCs, thereby reaching high transgene expression levels in 6-well plates with levels of around 100 ng/ml IL1RA. MSC aggregate cultures which were maintained in chondrogenic media without IL1ß supplementation revealed a chondrogenic phenotype by means of strong positive staining for collagen type II and matrix proteoglycan (Alcian blue). Addition of IL1ß was inhibitory to chondrogenesis in untreated control pellets. In contrast, foamyviral mediated IL1RA expression rescued the chondrogenesis in pellets cultured in the presence of IL1ß. Transduced MSC pellets reached thereby very high IL1RA transgene expression levels with a peak of 1087 ng/ml after day 7, followed by a decrease to 194 ng/ml after day 21, while IL1RA concentrations of controls were permanently below 200 pg/ml. Conclusion: Our results indicate that FVV are capable of efficient gene transfer to MSCs, while reaching IL1RA transgene expression levels, that were able to efficiently block the impacts of IL1ß in vitro. FVV merit further investigation as a means to study the potential as a gene transfer tool for MSC based therapies for cartilage repair.

18.
J Tissue Eng Regen Med ; 11(3): 877-886, 2017 03.
Article in English | MEDLINE | ID: mdl-25627968

ABSTRACT

Tissue-engineering techniques have been successful in developing cartilage-like tissues in vitro using cells from animal sources. The successful translation of these strategies to the clinic will likely require cell expansion to achieve sufficient cell numbers. Using a two-dimensional (2D) cell migration assay to first identify the passage at which chondrocytes exhibited their greatest chondrogenic potential, the objective of this study was to determine a more optimal culture medium for developing three-dimensional (3D) cartilage-like tissues using human cells. We evaluated combinations of commonly used growth factors that have been shown to promote chondrogenic growth and development. Human articular chondrocytes (AC) from osteoarthritic (OA) joints were cultured in 3D environments, either in pellets or encapsulated in agarose. The effect of growth factor supplementation was dependent on the environment, such that matrix deposition differed between the two culture systems. ACs in pellet culture were more responsive to bone morphogenetic protein (BMP2) alone or combinations containing BMP2 (i.e. BMP2 with PDGF or FGF). However, engineered cartilage development within agarose was better for constructs cultured with TGFß3. These results with agarose and pellet culture studies set the stage for the development of conditions appropriate for culturing 3D functional engineered cartilage for eventual use in human therapies. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Cartilage, Articular/physiology , Cell Movement , Chondrocytes/cytology , Tissue Engineering/methods , Cell Survival , DNA/metabolism , Humans , Middle Aged , Tissue Scaffolds/chemistry
19.
Stem Cell Res Ther ; 7(1): 154, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27765063

ABSTRACT

BACKGROUND: Lowering oxygen from atmospheric level (hyperoxia) to the physiological level (physioxia) of articular cartilage promotes mesenchymal stem cell (MSC) chondrogenesis. However, the literature is equivocal regarding the benefits of physioxic culture on preventing hypertrophy of MSC-derived chondrocytes. Articular cartilage progenitors (ACPs) undergo chondrogenic differentiation with reduced hypertrophy marker expression in hyperoxia but have not been studied in physioxia. This study sought to delineate the effects of physioxic culture on both cell types undergoing chondrogenesis. METHODS: MSCs were isolated from human bone marrow aspirates and ACP clones were isolated from healthy human cartilage. Cells were differentiated in pellet culture in physioxia (2 % oxygen) or hyperoxia (20 % oxygen) over 14 days. Chondrogenesis was characterized by biochemical assays and gene and protein expression analysis. RESULTS: MSC preparations and ACP clones of high intrinsic chondrogenicity (termed high-GAG) produced abundant matrix in hyperoxia and physioxia. Poorly chondrogenic cells (low-GAG) demonstrated a significant fold-change matrix increase in physioxia. Both high-GAG and low-GAG groups of MSCs and ACPs significantly upregulated chondrogenic genes; however, only high-GAG groups had a concomitant decrease in hypertrophy-related genes. High-GAG MSCs upregulated many common hypoxia-responsive genes in physioxia while low-GAG cells downregulated most of these genes. In physioxia, high-GAG MSCs and ACPs produced comparable type II collagen but less type I collagen than those in hyperoxia. Type X collagen was detectable in some ACP pellets in hyperoxia but reduced or absent in physioxia. In contrast, type X collagen was detectable in all MSC preparations in hyperoxia and physioxia. CONCLUSIONS: MSC preparations and ACP clones had a wide range of chondrogenicity between donors. Physioxia significantly enhanced the chondrogenic potential of both ACPs and MSCs compared with hyperoxia, but the magnitude of response was inversely related to intrinsic chondrogenic potential. Discrepancies in the literature regarding MSC hypertrophy in physioxia can be explained by the use of low numbers of preparations of variable chondrogenicity. Physioxic differentiation of MSC preparations of high chondrogenicity significantly decreased hypertrophy-related genes but still produced type X collagen protein. Highly chondrogenic ACP clones had significantly lower hypertrophic gene levels, and there was little to no type X collagen protein in physioxia, emphasizing the potential advantage of these cells.


Subject(s)
Chondrogenesis/physiology , Mesenchymal Stem Cells/metabolism , Oxygen/metabolism , Aged , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cartilage, Articular/metabolism , Cartilage, Articular/physiology , Cell Differentiation/physiology , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/physiology , Collagen Type I/metabolism , Collagen Type II/metabolism , Collagen Type X/metabolism , Female , Humans , Hypertrophy/metabolism , Hypertrophy/physiopathology , Male , Mesenchymal Stem Cells/physiology , Middle Aged
20.
Biotechnol Lett ; 38(11): 1967-1974, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27534541

ABSTRACT

OBJECTIVES: To compare in vitro chondrogenesis from bone marrow-derived mesenchymal stem cells using concave microwell plates with those obtained using culture tubes. RESULTS: Pellets cultured in concave microwell plates had a significantly higher level of GAG per DNA content and greater proteoglycan content than those cultured in tubes at day 7 and 14. Three chondrogenic markers, SOX-9, COL2A1 and aggrecan, showed significantly higher expression in pellets cultured in concave microwell plates than those cultured in tubes at day 7 and 14. At day 21, there was not a significant difference in the expression of these markers. COL10A1, the typical hypertrophy marker, was significantly lower in concave microwell plates during the whole culture period. Runx-2, a marker of hypertrophy and osteogenesis, was significantly lower at day 7 in pellets cultured in concave microwell plates than those cultured in tubes. CONCLUSION: Concave microwell plates provide a convenient and effective tool for the study of in vitro chondrogenesis and may replace the use of propylene culture tube.


Subject(s)
Cell Culture Techniques/instrumentation , Chondrogenesis , Mesenchymal Stem Cells/cytology , Aged , Cell Differentiation , Cells, Cultured , DNA/analysis , Gene Expression Regulation , Humans , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...